Discovery of a Novel Series of Imidazo[1,2-a]pyrimidine Derivatives as

Oct 19, 2015 - The active site was defined according to the crystal structure of Lp-PLA2 in complex with its substrate (PDB ID: 3D5E).(58) Water molec...
1 downloads 4 Views 910KB Size
Subscriber access provided by NATIONAL UNIV OF SINGAPORE

Article

Discovery of a Novel Series of Imidazo[1,2-a]pyrimidine Derivatives as Potent and Orally Bioavailable Lipoprotein-Associated Phospholipase A2 Inhibitors Xinde Chen, Wenwei Xu, Kai Wang, Mingguang Mo, Wei Zhang, Lili Du, Xiaojing Yuan, Yechun Xu, Yi-Ping Wang, and Jianhua Shen J. Med. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jmedchem.5b01024 • Publication Date (Web): 19 Oct 2015 Downloaded from http://pubs.acs.org on October 22, 2015

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Medicinal Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Discovery of a Novel Series of Imidazo[1,2-a]pyrimidine Derivatives as Potent and Orally Bioavailable Lipoprotein-Associated Phospholipase A2 Inhibitors Xinde Chen,† Wenwei Xu,† Kai Wang, Mingguang Mo, Wei Zhang, Lili Du, Xiaojing Yuan, Yechun Xu, Yiping Wang,* Jianhua Shen*

State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 58

ABSTRACT Inhibition of lipoprotein-associated phospholipase A2 (Lp-PLA2) has been suggested to be a promising therapeutic strategy for several inflammation-associated diseases, including atherosclerosis, Alzheimer’s disease, and diabetic macular edema. Herein, we report the discovery

of

a

novel

series

of

Lp-PLA2

inhibitors

constructed

on

an

imidazo[1,2-a]pyrimidine scaffold through a conformational restriction strategy. Structure-activity relationship (SAR) analysis resulted in identification of several compounds with high potency in vitro and good metabolic stability in liver S9 fractions. Compounds 7c and 14b selected for further exploration in vivo demonstrated excellent pharmacokinetic profiles and exhibited significant inhibitory efficacy in SD rats upon oral dosing.

ACS Paragon Plus Environment

Page 3 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

INTRODUCTION Lipoprotein-associated

phospholipase

A2

(Lp-PLA2)

previously

known

as

platelet-activating factor acetylhydrolase (PAF-AH), is a member of the phospholipase A2 superfamily.1 Lp-PLA2 is produced mainly by macrophages, monocytes, T-lymphocytes, and mast cells.2–4 In human plasma, Lp-PLA2 circulates mainly with low-density lipoprotein5 and can hydrolyze oxidized modified phosphatidylcholine to generate oxidized free fatty acids and lysophosphatidylcholine (lyso-PC),6 and both products have been shown to induce inflammatory responses.7, 8 Lyso-PC, for example, has been found to be involved in leukocyte activation, induction of apoptosis, and mediation of endothelial dysfunction.9, 10 Consistent with these results, Lp-PLA2 has been considered to be a promising therapeutic target for atherosclerosis.11–13 Very recently, it was reported that inhibition of Lp-PLA2 might protect against the damage to the blood-brain barrier caused by hyperglycemia and hypercholesterolemia, suggesting that inhibition of Lp-PLA2 could be used for treating neurodegenerative diseases.14 In addition, inhibition of Lp-PLA2 was recently reported to have beneficial effects against diabetic macular edema (DME),15, 16 a severe complication whose pathogenesis is directly related to the breakdown of the blood-retinal barrier.17, 18 To date, a number of Lp-PLA2 inhibitors have been reported, including azetidinones,19 oximes,20,

21

amides of xanthurenic acid,22,

23

carbamates,24 and

pyrimidones.25–39 Among them, several compounds have progressed into clinical trials (Figure 1), and darapladib has been the most rapidly developed.40 Although darapladib

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

was ineffective in two phase-III trials focusing on coronary heart diseases,41, 42 another phase-II trial revealed that oral administration of darapladib for 3 months modestly reduced macular edema and enhanced vision acuity in center-involved DME patients.16 Two other compounds, rilapladib (once also in phase-II trial for atherosclerosis43) and GSK2647544 (structure not disclosed), which are in phase-II and phase-I trials, respectively, are in development for the treatment of Alzheimer’s disease.

`

Figure 1. Chemical structures of Lp-PLA2 inhibitors in clinical trials. Previously, our research team reported a series of darapladib analogs with high potency in vitro.44, 45 However, high oral efficacy through further structural modifications was hard to obtain because of low plasma exposure. Likewise, darapladib as the initial compound also shows relatively low oral plasma exposure because of low oral bioavailability.31, 46 We postulated that the limited oral plasma exposure of darapladib and its analogs should be mainly due to their large molecular sizes and high lipophilicity (darapladib: molecular weight (MW), 666.78; cLogP, 8.33).47 Therefore, we sought to develop a new class of Lp-PLA2 inhibitors with lower MW and cLogP to obtain better pharmacokinetic properties, so that sufficient plasma concentrations of these compounds could be achieved to effectively inhibit Lp-PLA2 enzyme in plasma after oral administration.

ACS Paragon Plus Environment

Page 4 of 58

Page 5 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

In our ongoing exploration of novel molecular scaffolds of Lp-PLA2 inhibitors, we recently undertook rational drug design on compound 1 (Figure 2A), which was newly developed by GlaxoSmithKline (GSK) scientists, and reported to have high potency in vitro.32 Moreover, compound 1 provided a smaller molecular size as well as lower lipophilicity (MW, 502.88; cLogP, 4.48) compared with darapladib, and our preliminary pharmacokinetic experiments revealed that compound 1 demonstrated greater plasma exposure than darapladib when dosed orally to SD rats (Table 1). Nevertheless, besides these virtues, compound 1 showed limited elimination half-life in vivo (t1/2 = 2.08 h). Through further evaluation of metabolic stability in rat liver S9 fractions, we confirmed that the high clearance of compound 1 in vivo was mainly due to its poor metabolic stability (CLint = 74.4 mL/min/kg). Structural restriction has been reported to contribute to

greater

metabolic

stability,48–51

and

bicyclic

structures,

such

as

imidazo[1,2-a]pyrimidine, have been commonly used as a versatile building block in medicinal chemistry,52–54 so we were tempted to use a structural restriction strategy to replace the pyrimidone group in compound 1 with a new imidazo[1,2-a]pyrimidine scaffold (Figure 2A). Based on this design, the first-prepared compound (7a) demonstrated better metabolic stability in human and rat liver S9 fractions as compared with compound 1, and displayed good potency against recombinant human Lp-PLA2 (rhLp-PLA2). Encouraged by these results, we undertook systematic structure-activity relationship (SAR) explorations of imidazo[1,2-a]pyrimidine derivatives. Herein, we describe the discovery of imidazo[1,2-a]pyrimidine derivatives as a novel

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

class of Lp-PLA2 inhibitors. These compounds are potent in vitro, metabolically stable, and exhibit excellent pharmacokinetic profiles and high inhibitory efficacy in vivo. The synthesis and pharmacological evaluation are reported.

Figure 2. (A) Design of imidazo[1,2-a]pyrimidine derivatives as Lp-PLA2 inhibitors. (B) Systematic SAR explorations of imidazo[1,2-a]pyrimidine derivatives.

ACS Paragon Plus Environment

Page 6 of 58

Page 7 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Table 1. Pharmacokinetic parameters of compound 1 and darapladib after oral administration to SD ratsa

a

compd

AUC0-∞ (µg·h/mL)

t1/2 (h)

Cmax (µg/mL)

1b

3.38

2.08

0.78

darapladibc

2.11

11.50

0.15

n = 5 animals/group; badministered at 25 mg/kg; cadministered at 50 mg/kg. Data are the

mean. CHEMISTRY The synthesis of imidazo[1,2-a]pyrimidine derivatives 7a-e is summarized in Scheme 1. A

nucleophilic

substitution

reaction

between

benzyl

mercaptan

and

4-chloropyrimidin-2-amine in the presence of sodium hydride yielded intermediate 2, and subsequent cyclization of 2 provided intermediate 3.55 Direct arylation of 3 with 5-bromopyrimidine using palladium(II) acetate as a catalyst gave intermediate 4,56 which was oxidized using m-CPBA to provide 5 as a mixture of the sulphone and sulfoxide. A nucleophilic substitution reaction between 5 and hydroxyl or thiol derivatives 6a-e (6a-e: see Supporting Information for synthesis details) using sodium hydride as a base afforded compounds 7a-e.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 58

Scheme 1. Synthesis of analogs 7a-ea

a

Reagents and conditions: (a) 4-chloropyrimidin-2-amine, sodium hydride, THF, rt; (b)

2-bromoacetaldehyde diethyl acetal, 48% HBr aqueous solution, 80 °C, 6 h, then rt, NaHCO3, 2, 70 °C, 2 h; (c) 5-bromopyrimidine, Pd(OAc)2, KOAc, DMA, 140 °C; (d) m-CPBA, DCM, rt; (e) sodium hydride, THF, rt. Compounds 10a-d were synthesized starting from benzyl alcohol 6c and 4-chloropyrimidin-2-amine using a method similar to that described for intermediate 4 (Scheme 2). Compound 10e was obtained by an alternative route: bromination of 9 gave intermediate

11,

and

subsequent

(1-methyl-1H-pyrazol-4-yl)boronic

acid

Suzuki using

coupling

of

11

and

bis(triphenylphosphine)palladium(II)

dichloride as a catalyst gave the desired compound 10e. The synthesis of 10f was commenced by a cyclization reaction between 8 and 12 in ethanol.

ACS Paragon Plus Environment

Page 9 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Scheme 2. Synthesis of compounds 10a-fa

a

Reagents and conditions: (a) 4-chloropyrimidin-2-amine, sodium hydride, THF, rt; (b)

2-bromoacetaldehyde diethyl acetal, 48% HBr aqueous solution, 80 °C, 6 h, then rt, NaHCO3, 8, 70 °C, 2 h; (c) R1Br, Pd(OAc)2, KOAc, DMA, 140 °C; (d) NBS, THF, rt; (e) (1-methyl-1H-pyrazol-4-yl)boronic

acid,

PdCl2(PPh3)2,

Na2CO3,

dioxane,

H2O,

microwave, 150 °C, 30 min; (f) ethanol, H2O, reflux. As shown in Scheme 3, the biaryl analogs 14a-m were synthesized in a manner similar to that described for compounds 7a-e starting from 5 and the corresponding benzyl alcohol 13a-m (13a-m: see Supporting Information for synthesis details).

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Scheme 3. Synthesis of biaryl analogs 14a-ma

a

Reagents and conditions: (a) sodium hydride, THF, rt.

RESULTS AND DISCUSSION SAR in vitro. Compounds were first evaluated for their inhibitory activity against rhLp-PLA2 at two concentrations (100 nM and 10 nM) in vitro, and rhLp-PLA2 activity was measured using 2-thio-PAF as the substrate. IC50 values against rhLp-PLA2 for those compounds with an acceptable inhibitory activity (>30% at 10 nM) were tested to ascertain their inhibitory potency precisely.

ACS Paragon Plus Environment

Page 10 of 58

Page 11 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

SAR analyses mainly focused on the influences of three regions on rhLp-PLA2 inhibitory activity (Figure 2B): the linker between the imidazo[1,2-a]pyrimidine moiety and diphenyl ether moiety; substituents at the 3-position of imidazo[1,2-a]pyrimidine (R1); diphenyl ether moiety. Effects of the linker were first explored (Table 2). Compound 7b with the oxygen atom directly connected to the diphenyl ether moiety showed a complete loss of potency, and elongation of the linker to three carbon atoms (7d) was also not tolerated. Compound 7c with a single methylene spacer demonstrated comparable potency with the initial hit compound 7a. Replacement of the oxygen atom with a sulfur atom (7e) reduced potency markedly. These results suggested that rhLp-PLA2 inhibitory activity was affected considerably by the length of the linker and linking atom.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 58

Table 2. SAR of the linker

% inhibition against compd

X

IC50 (nM)

rhLp-PLA2

n

100 nM

10 nM 55

11.1

55

10.6

7a

O

2

87

7b

O

0

0

7c

O

1

89

7d

O

3

19

7e

S

1

28

1

98

78

3.4

darapladib

92

88

0.7a

a

Reported IC50 = 0.25 nM. On the basis of the high inhibitory activity, 7c was selected for further optimization.

Our next objective was to investigate the influences of R1 on potency, and representative compounds are shown in Table 3. Among the modifications, we found that removal of the pyrimidine ring resulted in a loss of potency (9). Replacement of the pyrimidine ring with a p-methoxy-phenyl ring was not well tolerated (10a), whereas the pyridine derivatives 10b and 10c restored potency to a certain degree. These SARs suggested that an aromatic

ACS Paragon Plus Environment

Page 13 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

ring and hydrogen-bond acceptor(s) were required at this moiety in the molecule. This hypothesis was further confirmed by the 2-methoxy-pyrimidine compound 10d and 1-methyl-pyrazole compound 10e: both compounds were more potent than the p-methoxy-phenyl compound 10a. Loss of potency after relocation of the pyrimidine ring to the 2-position (10f) suggested that substitution at the 3-position was favored for potency. Table 3. Influences of R1 on the inhibitory activity of rhLp-PLA2

% inhibition against compd

R1

R2

IC50 (nM)

rhLp-PLA2 100 nM

10 nM

H

89

55

H

26

10a

H

58

25

10b

H

76

34

19.3

10c

H

78

32

16.0

10d

H

79

44

13.4

10e

H

78

32

16.9

7c 9

10f

H

H

12

ACS Paragon Plus Environment

10.6

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Finally, we explored the effects of the diphenyl ether moiety. As shown in Table 4, we first investigated the influences of the central benzene ring. Introduction of a methyl substituent ortho to the diphenyl ether linkage resulted in a decrease in potency (14a). In contrast, introduction of a small, electron-withdrawing fluorine group (14b) or cyano group (14c) increased the activity as compared to 7c. Encouraged by the results of 14b and 14c, we then turned our attention to the introduction of electron-withdrawing groups in the central benzene ring. However, compounds with a bulkier substituent such as a chloro group (14d) or trifluoromethyl group (14e) were less potent. In addition, further incorporation of a fluorine group in the fluorine-substituted derivative 14b maintained inhibitory potency (14f). However, replacement of the central benzene ring with a pyridine ring or five-membered heteroaromatic ring provided a lower cLogP value was not tolerated (data not shown). These SARs led us to speculate that slight electron deficiency and hydrophobicity of the central aromatic ring was beneficial for the binding to the Lp-PLA2 enzyme. Next, keeping the di-fluorine benzene ring constant, we examined the effects of the terminal benzene ring. Representative compounds are shown in Table 4. A benzene ring without substituents (14g) was less active than compound 14f. Replacement of the chlorine atom with a fluorine atom (14h) or removal of the chlorine atom (14i) resulted in compounds with slightly decreased potency. Replacement of the trifluoromethyl group with a chlorine atom (14j) also resulted in a slight decrease in potency, and migration of the trifluoromethyl group to the para position (14k) was poorly tolerated. As with the central benzene ring, alteration of the terminal benzene ring to the

ACS Paragon Plus Environment

Page 14 of 58

Page 15 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

pyridine ring also resulted in low-activity compounds (14l and 14m). Thus, we speculated that the terminal benzene ring played a key role in hydrophobic interactions with Lp-PLA2 enzyme, and Lp-PLA2 inhibitory activity was obviously affected by the substituents on the terminal benzene ring.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 58

Table 4. Effects of the diphenyl ether on the inhibitory activity of rhLp-PLA2

% inhibition against IC50 (nM)

rhLp-PLA2

compd

100 nM

10 nM

7c

89

55

14a

70

15

14b

95

78

3.7

14c

97

68

2.9

14d

74

11

14e

3

10.6

F

14f

100

O

82

2.3

F

14g

47

14h

96

66

6.5

14i

91

47

10.9

14j

94

57

6.3

14k

79

29

14l

57

8

14m

21

To

further

gain

insight

into

the

binding

ACS Paragon Plus Environment

interactions

of

these

Page 17 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

imidazo[1,2-a]pyrimidine derivatives with Lp-PLA2 enzyme, docking studies were then performed to investigate the docking pose of compound 14b into Lp-PLA2 (Figure 3). Key interactions between this compound and Lp-PLA2 are hydrogen bonds and several hydrophobic interactions. Q352 formed two hydrogen bonds with 14b. The pyrimidine ring in position R1 formed an edge-to-face π-π stacking interaction with W298. In the case of imidazo[1,2-a]pyrimidine ring, the pyrimidine moiety formed hydrophobic interactions with H351 and L153. The central and terminal benzene rings are participated in a number of hydrophobic interactions, most notably to F110, G154, Y160, and F357.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 3. Docking pose of compound 14b showing key interactions with Lp-PLA2 binding site. Carbon, oxygen, nitrogen, fluorine and chlorine atoms are colored cyan, red, blue, limon, and green, respectively. Compound 14b is represented as a ball-and-stick model. The protein is shown as light grey cartoons. (A) The binding pocket is shown by molecular surface. (B) Interactions between 14b and Lp-PLA2. Hydrogen bonds are depicted as dotted lines. Key residues at the binding site are shown as sticks. To account for any non-specific binding effects in plasma, selected compounds with relatively good activity against rhLp-PLA2 were further evaluated for their potency against Lp-PLA2 in human plasma. As shown in Table 5, potency against Lp-PLA2 in human plasma correlated well with potency against rhLp-PLA2. For example, compounds 14c and 14f act as the most potent compounds against rhLp-PLA2, displayed rather high potency against Lp-PLA2 in human plasma, and potency against rhLp-PLA2 of 14c and

ACS Paragon Plus Environment

Page 18 of 58

Page 19 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

14f (14c, 68% at 10 nM; 14f, 82% at 10 nM) were similar to their potency against Lp-PLA2 in human plasma (14c, 83% at 10 nM; 14f, 87% at 10 nM). These findings suggested that non-specific binding had a negligible effect on the binding affinity between these imidazo[1,2-a]pyrimidine derivatives and Lp-PLA2 enzyme in human plasma. Before we evaluated efficacy in vivo, we also assessed the activity of selected compounds against Lp-PLA2 in rat and mouse plasma (Table 5). Surprisingly, potency in the plasma of rats and mice differed from the potency observed in human plasma for these imidazo[1,2-a]pyrimidine derivatives. For instance, compound 14f displayed a high activity against Lp-PLA2 in human plasma (87% at 10 nM), but showed a relatively low activity in rat plasma (26% at 100 nM). In addition, in mouse plasma, all of these imidazo[1,2-a]pyrimidine derivatives showed quite low potency against the Lp-PLA2 enzyme. Despite this species difference, most of these imidazo[1,2-a]pyrimidine derivatives provided >50% inhibition of Lp-PLA2 in rat plasma at 100 nM, which were better than that observed for compound 1.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 58

Table 5. Inhibitory activity against Lp-PLA2 in human plasma, rat plasma, and mouse plasma % inhibition in plasma human

compd

rat

100 nM

10 nM

100 nM

7a

87

56

49

7c

91

58

76

10e

85

42

14b

84

14c

mouse 10 nM

1 µM

100 nM

53

2

10

56

10

64

13

28

60

70

11

13

94

83

68

14

0

14f

96

87

26

8

1

96

86

48

91

18

darapladib

100

93

98

99

96

84

Metabolic stability tests in liver S9 fractions. Compounds with relatively good activity against rhLp-PLA2 were also selected to evaluate their metabolic stability in human and rat liver S9 fractions (Table 6). Compound 7c with a single methylene spacer displayed much better metabolic stability compared with the hit compound 7a. Replacement of the pyrimidine ring with 1-methyl-pyrazole (10e) also resulted in good metabolic stability. Compounds with different substituents in the central benzene ring demonstrated different metabolic stability. The cyano-substituted derivative 14c showed relatively poor

ACS Paragon Plus Environment

Page 21 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

metabolic stability, whereas the fluorine-substituted derivatives 14b and 14f were quite stable in human and rat liver S9 fractions. Table 6. Metabolic stability in liver S9 fractions of selected compounds liver S9 stability (t1/2, min) compd

a

human

rat

7a

42

110

7c

116

348

10e

180

NCa

14b

331

169

14c

106

40

14f

212

196

1

33

51

NC = cannot be calculated. The half-life was too long to be calculated in this system.

Pharmacokinetic evaluation. For assessment of the pharmacokinetic properties of these imidazo[1,2-a]pyrimidine derivatives in vivo, compounds 7c, 14b, and 14c were selected for evaluation in male SD rats through oral administration. The concentration-time curves are shown in Figure 4 (with the data of compound 1 and darapladib included for comparison), and the key pharmacokinetic parameters are summarized in Table 7. Pharmacokinetic studies showed that the in vivo half-lives of these compounds correlated well with the metabolic stability in rat liver S9 fractions. With good metabolic stability in rat liver S9 fractions, in vivo half-lives of 7c and 14b were 9.44 h and 13.34 h,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 58

respectively, much longer than that of compound 1 (2.08 h). Compound 14c was less stable in rat liver S9 fractions, and displayed a shorter in vivo half-life (3.24 h). Furthermore, 7c, 14b, and 14c achieved a maximum concentration (Cmax) of 3.52, 2.60 and 1.40 µg/mL, respectively, whereas the Cmax of compound 1 was 0.78 µg/mL. Consequently, AUC values for 7c (88.05 µg·h/mL), 14b (78.14 µg·h/mL), and 14c (16.13 µg·h/mL) were higher than that for compound 1 (3.38 µg·h/mL). Taken together, these data

suggested

that

imidazo[1,2-a]pyrimidine

derivatives

possess

promising

pharmacokinetic profiles in vivo. Table 7. Pharmacokinetic parameters of selected compounds after oral administration to SD ratsa

a

compd

AUC0-∞ (µg·h/mL)

t1/2 (h)

Cmax (µg/mL)

7c

88.05

9.44

3.52

14b

78.14

13.34

2.60

14c

16.13

3.24

1.40

n = 5 animals/group, administered at 25 mg/kg. Data are the mean.

ACS Paragon Plus Environment

Page 23 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Figure 4. Concentration-time curves of selected compounds after oral administration to SD rats (n=5). Compounds 7c, 14b, 14c and 1 were administrated at 25 mg/kg, darapladib was administered at 50 mg/kg. Assessment of the inhibitory activity of Lp-PLA2 in vivo. Based on their very promising profiles, compounds 7c and 14b were selected for evaluating inhibitory activity in male SD rats through oral administration at a single dose of 25 mg/kg, with the data for compound 1 and darapladib included for comparison (Figure 5). The relatively low inhibitory activity of compound 1 in vivo could be due to its modest pharmacokinetic profile and relatively low potency against Lp-PLA2 in rat plasma in vitro. With much better pharmacokinetic properties and slightly greater potency against Lp-PLA2 in rat plasma in vitro, 7c and 14b exhibited much better inhibitory activity in vivo when compared with compound 1. Both compounds produced ≈75% inhibition of Lp-PLA2 activity after 5 h and could effectively inhibit Lp-PLA2 activity over 24 h.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 58

Figure 5. Relative activity of Lp-PLA2 in the serum of SD rats after a single oral dose of 25 mg/kg (n = 5). CONCLUSION Among reported Lp-PLA2 inhibitors, darapladib has been the most rapidly developed compound. However, darapladib demonstrated low oral bioavailability due to its physicochemical properties, such as high values of MW (666.78) and cLogP (8.33). In an effort to find potent and orally bioavailable Lp-PLA2 inhibitors, we focused on the development of compounds with lower values of MW and cLogP. In this work, we chose compound 1 (MW, 502.88; cLogP, 4.48) from GSK as a starting point. In view of the poor metabolic stability of compound 1, structural modification using conformational restriction yielded imidazo[1,2-a]pyrimidine as a novel

scaffold.

SAR

explorations

resulted

in

identification

of

several

imidazo[1,2-a]pyrimidine derivatives with good inhibitory activity in vitro and favorable metabolic stability in liver S9 fractions. Several compounds that proceeded to pharmacokinetic evaluation in vivo showed much better pharmacokinetic profiles

ACS Paragon Plus Environment

Page 25 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

compared with those observed for darapladib and compound 1. Compounds 7c and 14b selected for efficacy assessment in vivo demonstrated robust inhibitory potency in male SD rats at a single oral dose of 25 mg/kg, this activity was comparable with that for darapladib and much better than that for compound 1. Taken together, results generated so far suggest that imidazo[1,2-a]pyrimidine derivatives with reasonable physicochemical properties, are potent, and orally bioavailable Lp-PLA2 inhibitors. In this way, they can serve as valuable probes to further study the role of Lp-PLA2 in vivo across various inflammation-associated diseases. We also observed a species difference across humans, rats and mice. Additional studies on identification of compounds to overcome this species difference are underway. EXPERIMENTAL SECTION In vitro assay to measure the inhibitory activity of Lp-PLA2.57 Activities against recombinant human Lp-PLA2 (rhLp-PLA2), human plasma, rat plasma and mouse plasma Lp-PLA2 were measured using 2-thio-PAF as the substrate. Briefly, 10 µL of the rhLp-PLA2 enzyme (or plasma) and 10 µL of a DMSO solution of the compound were added to 0.1 mol/L Tris-HCl (pH 7.2) containing 1 mmol/L EGTA, 50 µmol/L 2-thio-PAF and 10 µL of 2 mmol/L 5,5′-dithiobis (2-nitrobenzoic acid) in a total volume of 200 µL. The assay was carried out using a plate reader to obtain absorbance values at 414 nm every minute for 10 min. Percent inhibition was determined using the following equation: Inhibition % = 1 −

Vmax − Vmax × 100% Vmax !" − Vmax

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Vmax: slope of absorbance values for 10 min, calculated by MolecularDevice, SpectraMax M2e. The blank sample contained no rhLp-PLA2 enzyme (or plasma) or test compound in assay buffer. The positive sample contained no test compound. This study has been approved and supervised by Institutional Animal Care and Use Committee (IACUC), Shanghai Institute of Materia Medica, Chinese Academy of Sciences (IACUC Approval Number: SIMM-2014-08-WYP-18) and the Ethics Committee of Shanghai Xuhui Central Hospital. Metabolic stability tests in human and rat liver S9 fractions. The test compound was dissolved in DMSO and diluted to the desired concentration with an aqueous solution of 0.1% BSA. Liver S9 (0.33 mg/mL; pooled human liver S9 purchased from Celsis In Vitro Technologies; Wistar rat liver S9 purchased from Research Institute for Liver Diseases), test compounds (0.1 µM), MgCl2 (5.0 mM), BSA (0.005%), and NADPH (1.0 mM) in Tris buffer (0.1 M, pH 7.4) were incubated in a 96-well plate at 37 °C. An aliquot was removed at each time point and the enzymatic reaction stopped by protein precipitation in cold methanol. Half-lives of the compounds in liver S9 fractions were calculated based on the first-order rate constants that were measured. Intrinsic clearance was calculated using the following equation: CL =

0.693 mL of incubation mg of liver S9 protein g of tissue × × × +, -+./0 t1/3 mg of liver S9 protein g of tissue kg of weight

Scaling factors for the human liver (145-mg S9 protein/g liver and 24.3-g liver/kg body weight) and rat liver (179-mg S9 protein/g liver and 40-g liver/kg body weight) were employed in this calculation.

ACS Paragon Plus Environment

Page 26 of 58

Page 27 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Docking protocols. A crystal structure of Lp-PLA2 (PDB ID: 3D59) was used as the receptor structure. The active site was defined according to the crystal structure of Lp-PLA2 in complex with its substrate (PDB ID: 3D5E).58 Water molecules and ions were deleted. Receptor was prepared using Protein Preparation and Grid Preparation tools in the Schrödinger Maestro interface. The 3D structures of substrate were generated with the Schrödinger program LigPrep. Automated docking was performed with Glide 5.5.59 Standard Precision (SP) calculations were carried out with default settings. The OPLS-2005 force field was used for minimization and grid generation, while OPLS-2001 force field was used for docking. The docked conformation of the compound with the lowest energy was selected for study. Animals. Male Sprague−Dawley (SD) rats were obtained from Shanghai SLAC Laboratory Animal Co. Ltd. (Shanghai, China). Animal experiments were approved by Animal Care and Use Committee, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Pharmacokinetic studies. Test compounds (0.5% carboxymethylcellulose sodium) were subjected to pharmacokinetic studies on male SD rats with five animals in each group. Test compounds were administered via the oral route by gavage at 25 mg/kg. Serial specimens were collected pre-dose as well as 1, 3, 5, 7, 24 and 48 h after administration and quantified by liquid chromatography-mass spectrometry. Pharmacokinetic parameters were calculated from the mean serum concentration by non-compartmental analyses using DAS software 2.1.1.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 58

Assay to measure the inhibitory activity of Lp-PLA2 in vivo. A group of five male SD rats were fasted overnight, and administered test compounds by gavage at 25 mg/kg. Test compounds were formulated in 0.5% carboxymethylcellulose sodium. Blood samples were drawn pre-dose as well as 1, 3, 5, 7 and 24 h after administration to measure Lp-PLA2 activity in serum. Lp-PLA2 activity in the serum of SD rats was measured according to the method described for measurement of Lp-PLA2 inhibitory activity in vitro. Materials and methods. All reagents were purchased from commercial suppliers and used without further purification unless otherwise stated. Yields were not optimized. Microwave reactions were performed in a Biotage Initiator. 1H NMR and

13

C NMR

spectra were recorded on a Varian-Mercucy Plus-300 or a Bruker AC400 or a Bruker AC500 NMR spectrometer using tetramethylsilane as an internal reference. Low-resolution mass spectra were determined on Agilent liquid-chromatography mass spectrometer system that consisted of an Agilent 1260 infinity LC coupled to Agilent 6120

Quadrupole

mass

spectrometer

(electrospray

positive

ionization;

ESI).

High-resolution mass spectra were conducted on a triple TOF 5600+ MS/MS system (AB Sciex, Concord, Ontario, Canada) in the positive ESI mode. The purity of test compounds was determined by HPLC (Agilent ChemStation, Agilent Eclipse XDB-C18, 5 µM, 4.6×150 mm, 30 °C, UV 240 nM, flow rate = 1.0 mL/min) with aqueous CH3CN (50-90%) containing ammonium formate (10 mmol/L) for 25 min. All the assayed compounds possess ≥95% purity. Column chromatography was performed on silica gel

ACS Paragon Plus Environment

Page 29 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

(200-300 mesh), preparative TLC was performed on HSGF 254 (0.4-0.5 mm thickness; Yantai Jiangyou Company, Yantai, Shangdong, China). 4-(benzylthio)pyrimidin-2-amine

(2).

To

a

suspension

of

4-chloropyrimidin-2-amine (2.00 g, 15.44 mmol) and sodium hydride (1.85 g, 46.25 mmol, 60% in mineral oil) in THF (50 mL) at 0 °C,was added benzyl mercaptan (1.60 mL, 13.65 mmol) dropwise over a period of 0.5 h, the reaction mixture was stirred at rt overnight. Then the reaction was quenched with NH4Cl solution, diluted with ethyl acetate (50 mL). The organic layer was separated, washed with brine, part of the organic solvent was evaporated under reduced pressure, the precipitates formed were collected by filtration to give 2 as a white solid (1.80 g, 54%).1H NMR (400 MHz, Chloroform-d) δ 7.85 (d, J = 5.5 Hz, 1H), 7.41 (d, J = 7.5 Hz, 2H), 7.32−7.18 (m, 3H), 6.50 (d, J = 5.5 Hz, 1H), 4.40 (s, 2H). MS (ESI): m/z 218 [M+H]+. 7-(benzylthio)imidazo[1,2-a]pyrimidine (3). A solution of 2-bromoacetaldehyde diethyl acetal (7.80 mL, 66.00 mmol) and HBr (2.70 mL, 24.02 mmol, 48% aqueous solution) in 95% ethanol (40 mL) was heated at 80 °C for 6 h. After the solution was cooled down to rt, solid NaHCO3 (3.50 g, 41.66 mmol) was added in small portions, follow by 2 (1.80 g, 8.29 mmol), the mixture was further stirred at 70 °C for 3 h. After the reaction was complete, the solvent was evaporated under reduced pressure, DCM (50 mL) was added to the residue, dried over MgSO4, filtered, and concentrated. The residue was purified with column chromatography (DCM/MeOH = 20:1) to yield 3 (684 mg, 34%) as

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

a yellow solid. 1H NMR (400 MHz, Chloroform-d) δ 8.10 (d, J = 7.0 Hz, 1H), 7.62 (s, 1H), 7.55−7.28 (m, 6H), 6.66 (d, J = 7.0 Hz, 1H), 4.57 (s, 2H). MS (ESI): m/z 242 [M+H]+. 7-(benzylthio)-3-(pyrimidin-5-yl)imidazo[1,2-a]pyrimidine (4). To a solution of 3 (684 mg, 2.84 mmol), 5-bromopyrimidine (496 mg, 3.12 mmol) in DMA (15 mL), was added Pd(OAc)2 (64 mg, 0.28 mmol), KOAc (556 mg, 5.67 mmol), the mixture was heated at 140 °C under nitrogen for 2 h. After the solution was cooled down, diluted with water (50 mL), and extracted with DCM (50 mL × 3), the organic layers were combined, washed with brine, dried over Na2SO4, filtered, and concentrated. The residue was purified with column chromatography (DCM/MeOH = 20:1) to yield 4 (370 mg, 41%) as a yellow solid. 1H NMR (300 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.93 (s, 2H), 8.22 (d, J = 7.2 Hz, 1H), 7.83 (s, 1H), 7.49 (d, J = 7.3 Hz, 2H), 7.39−7.28 (m, 3H), 6.78 (d, J = 7.5 Hz, 1H), 4.62 (s, 2H). MS (ESI): m/z 320 [M+H]+. Intermediate 5. To a solution of 4 (370 mg, 1.16 mmol) in DCM (60 mL) was added m-CPBA (715 mg, 2.90 mmol, 70% purity) at 0 °C, the mixture was stirred at 0 °C for 3 h. Then the reaction was quenched with sodium bicarbonate solution, the organic layer was separated, and washed with sodium bicarbonate solution, dried over Na2SO4, filtered, and concentrated. Intermediate 5 (240 mg) was obtained as a mixture, and can be used in next step without purification. MS (ESI): m/z 352[M+H]+, m/z 336[M+H]+. General Synthetic Procedure for 7a-e. To a solution of 5 (0.10 mmol) and

ACS Paragon Plus Environment

Page 30 of 58

Page 31 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

corresponding alcohol (0.10 mmol) in THF (2 mL) was added sodium hydride (0.25 mmol) at 0 °C. The reaction was stirred at rt for 1 h, and then quenched with NH4Cl solution, extracted with ethyl acetate (5 mL × 3), washed with brine, dried over Na2SO4, filtered, and concentrated. The residue obtained was purified with preparative TLC (DCM/MeOH = 30:1) to yield the desired compound. 7-(4-(4-chloro-3-(trifluoromethyl)phenoxy)phenethoxy)-3-(pyrimidin-5-yl)imida zo[1,2-a]pyrimidine (7a). The title compound was obtained as a white solid from 5 and 6a according to the general procedure in 72% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.26 (s, 1H), 8.92 (s, 2H), 8.30 (d, J = 7.4 Hz, 1H), 7.72 (s, 1H), 7.42 (d, J = 8.8 Hz, 1H), 7.32 (m, 3H), 7.06 (d, J = 9.0 Hz, 1H), 6.98 (d, J = 8.0 Hz, 2H), 6.54 (d, J = 8.3 Hz, 1H), 4.74 (t, J = 6.5 Hz, 2H), 3.16 (t, J = 6.4 Hz, 2H).

13

C NMR (126 MHz,

Chloroform-d) δ 162.83, 157.99, 156.30, 155.05 (2C), 154.49, 149.72, 134.12, 133.05, 132.63, 132.26, 130.65 (2C), 129.51 (q, J = 31.9 Hz), 125.55, 123.71, 123.40 (q, J = 273.9 Hz), 122.11, 119.56 (2C), 117.49 (q, J = 5.3 Hz), 117.25, 102.62, 67.68, 34.25. HRMS (ESI): m/z [M+H]+ calculated for C25H18ClF3N5O2: 512.1096, found: 512.1111. 7-(4-(4-chloro-3-(trifluoromethyl)phenoxy)phenoxy)-3-(pyrimidin-5-yl)imidazo[ 1,2-a]pyrimidine (7b). The title compound was obtained as a white solid from 5 and 6b according to the general procedure in 65% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.29 (s, 1H), 8.95 (s, 2H), 8.45 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.47 (d, J = 8.8 Hz, 1H), 7.41 (d, J = 2.7 Hz, 1H), 7.29 (d, J = 8.9 Hz, 2H), 7.14 (dd, J = 8.8, 2.7 Hz, 1H), 7.08 (d,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 58

J = 8.9 Hz, 2H), 6.81 (d, J = 7.3 Hz, 1H). 13C NMR (126 MHz, Chloroform-d) δ 162.24, 158.12, 156.11, 155.12 (2C), 153.37, 149.06, 148.26, 133.83, 133.08, 132.75, 129.65 (q, J = 31.4 Hz), 125.90, 123.63, 123.41 (2C), 122.38 (q, J = 274.0 Hz), 122.18, 120.40 (2C), 117.87 (q, J = 5.3 Hz), 117.53, 102.26. HRMS (ESI): m/z [M+H]+ calculated for C23H14ClF3N5O2: 484.0783, found: 484.0786. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyrimidin-5-yl)imida zo[1,2-a]pyrimidine (7c). The title compound was obtained as a white solid from 5 and 6c according to the general procedure in 75% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.93 (s, 2H), 8.33 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.54 (d, J = 8.6 Hz, 2H), 7.45 (d, J = 8.8 Hz, 1H), 7.34 (d, J = 2.9 Hz, 1H), 7.10 (dd, J = 8.8, 2.8 Hz, 1H), 7.04 (d, J = 8.6 Hz, 2H), 6.61 (d, J = 7.4 Hz, 1H), 5.55 (s, 2H).

13

C NMR (126 MHz,

Chloroform-d) δ 162.47, 157.94, 156.09, 155.80, 155.03 (2C), 149.61, 133.26, 132.73, 132.42, 131.81, 130.72 (2C), 129.62 (q, J = 31.8 Hz), 126.00, 123.78, 122.53, 122.35 (q, J = 273.9 Hz), 119.31 (2C), 117.89 (q, J = 5.3 Hz), 117.31, 102.49, 68.34. HRMS (ESI): m/z [M+H]+ calculated for C24H16ClF3N5O2: 498.0939, found: 498.0945. 7-(3-(4-(4-chloro-3-(trifluoromethyl)phenoxy)phenyl)propoxy)-3-(pyrimidin-5-yl )imidazo[1,2-a]pyrimidine (7d). The title compound was obtained as a yellow solid from 5 and 6d according to the general procedure in 35% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.26 (s, 1H), 8.93 (s, 2H), 8.31 (d, J = 7.4 Hz, 1H), 7.72 (s, 1H), 7.42 (d, J = 8.8 Hz, 1H), 7.31 (s, 1H), 7.24 (d, J = 8.5 Hz, 2H), 7.05 (dd, J = 8.8, 2.8 Hz, 1H),

ACS Paragon Plus Environment

Page 33 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

6.95 (d, J = 8.5 Hz, 2H), 6.56 (d, J = 7.4 Hz, 1H), 4.54 (t, J = 6.5 Hz, 2H), 2.81 (t, J = 8.4 Hz, 2H), 2.21−2.13 (m, 2H).

13

C NMR (126 MHz, Chloroform-d) δ 162.89, 157.88,

156.45, 154.99 (2C), 153.97, 149.87, 137.54, 133.16, 132.60, 132.17, 130.06 (2C), 129.46 (q, J = 31.9 Hz), 125.40, 123.85, 122.42 (q, J = 274.0 Hz), 121.98, 119.56 (2C), 117.41 (q, J = 5.5 Hz), 117.17, 102.52, 66.60, 31.48, 30.17. HRMS (ESI): m/z [M+H]+ calculated for C26H20ClF3N5O2: 526.1252, found: 526.1269. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)thio)-3-(pyrimidin-5-yl)imida zo[1,2-a]pyrimidine (7e). The title compound was obtained as a yellow solid from 5 and 6e according to the general procedure in 38% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.93 (s, 2H), 8.24 (d, J = 7.2 Hz, 1H), 7.83 (s, 1H), 7.51 (d, J = 8.6 Hz, 2H), 7.42 (d, J = 8.8 Hz, 1H), 7.32 (d, J = 2.9 Hz, 1H), 7.06 (dd, J = 8.8, 2.8 Hz, 1H), 6.96 (d, J = 8.6 Hz, 2H), 6.80 (d, J = 7.2 Hz, 1H), 4.61 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 161.93, 158.11, 155.92, 155.26, 154.13 (2C), 149.98, 133.94, 133.05, 132.67, 131.09 (2C), 129.73, 129.07, 125.72, 123.57, 122.93 (q, J = 298.6 Hz), 122.38, 119.38 (2C), 117.83 (q, J = 6.3 Hz), 117.44, 109.44, 33.46. HRMS (ESI): m/z [M+H]+ calculated for C24H16ClF3N5OS: 514.0711, found: 514.0705. 4-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)pyrimidin-2-amine

(8).

To a solution of 6c (3.00 g, 9.91 mmol) in anhydrous THF (100 mL) was added sodium hydride (0.80 g, 27.73 mmol, 60% in mineral oil) at 0 °C, 4-chloropyrimidin-2-amine (0.85 g, 6.56 mmol) was added after the mixture was stirred at 0 °C for 0.5 h, the mixture

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

was stirred at rt overnight. Then the reaction was quenched with NH4Cl solution, extracted with ethyl acetate (100 mL × 3), washed with brine, dried over NaSO4, filtered and concentrated, the residue obtained was purified with column chromatography (petroleum ether/ethyl acetate = 2:1) to yield intermediate 8 as a white solid (2.40 g, 93%). 1H NMR (400 MHz, Chloroform-d) δ 8.04 (d, J = 5.7 Hz, 1H), 7.44 (d, J = 8.6 Hz, 3H), 7.34 (d, J = 2.9 Hz, 1H), 7.09 (dd, J = 8.8, 2.9 Hz, 1H), 7.02 (d, J = 8.6 Hz, 2H), 6.15 (d, J = 5.7 Hz, 1H), 5.32 (s, 2H), 4.93 (s, 2H). MS (ESI): m/z 396 [M+H]+. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)imidazo[1,2-a]pyrimidin e (9). The title compound was obtained as a yellow solid from 8 using a method similar to that described for intermediate 3 in 34% yield. 1H NMR (300 MHz, Chloroform-d) δ 8.19 (d, J = 7.3 Hz, 1H), 7.56−7.49 (m, 3H), 7.44 (d, J = 8.7 Hz, 1H), 7.36−7.32 (m, 2H), 7.08 (dd, J = 8.8, 2.9 Hz, 1H), 7.03 (d, J = 8.6 Hz, 2H), 6.47 (d, J = 7.2 Hz, 1H), 5.51 (s, 2H). 13

C NMR (126 MHz, Chloroform-d) δ 162.08, 159.40, 155.90, 147.91, 134.93, 133.10,

132.70, 132.26, 130.63 (2C), 129.60 (q, J = 32.8 Hz), 125.89, 122.44, 122.37 (q, J = 274.68 Hz), 119.30 (2C), 117.86 (q, J = 5.0 Hz), 110.24, 101.33, 67.90. HRMS (ESI): m/z [M+H]+ calculated for C20H14ClF3N3O2: 420.0721, found: 420.0748. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(4-methoxyphenyl)imi dazo[1,2-a]pyrimidine (10a). The title compound was obtained as a white solid from 9 and 1-bromo-4-methoxybenzene using a method similar to that described for intermediate 4 in 61% yield. 1H NMR (400 MHz, Chloroform-d) δ 8.32 (d, J = 7.4 Hz, 1H), 7.54 (d, J

ACS Paragon Plus Environment

Page 34 of 58

Page 35 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

= 8.6 Hz, 2H), 7.51 (s, 1H), 7.42 (dd, J = 11.0, 8.8 Hz, 3H), 7.34 (d, J = 2.9 Hz, 1H), 7.08 (dd, J = 8.7, 2.8 Hz, 1H), 7.03 (dd, J = 8.6, 1.8 Hz, 4H), 6.47 (d, J = 7.4 Hz, 1H), 5.53 (s, 2H), 3.87 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 161.72, 159.75, 155.90, 148.03, 132.99, 132.70, 132.33, 130.65 (2C), 130.52, 129.60 (q, J = 31.9 Hz), 129.41 (2C), 125.89, 123.90, 122.43, 122.37 (q, J = 274.0 Hz), 120.90, 119.31 (2C), 117.86 (q, J = 5.4 Hz), 114.80 (2C), 113.23, 101.15, 67.92, 55.43. HRMS (ESI): m/z [M+H]+ calculated for C27H20ClF3N3O3: 526.1140, found: 526.1168. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyridin-3-yl)imidazo[ 1,2-a]pyrimidine (10b). The title compound was obtained as a white solid from 9 and 3-bromopyridine using a method similar to that described for intermediate 4 in 53% yield. 1

H NMR (400 MHz, Chloroform-d) δ 8.80 (d, J =1.2 Hz, 2H), 8.68 (dd, J = 4.4 Hz, 1.2

Hz, 2H), 8.35 (d, J = 7.4 Hz, 1H), 7.82 (dt, J = 7.8, 1.8 Hz, 1H), 7.71 (s, 1H), 7.54 (d, J = 8.5 Hz, 2H), 7.49−7.41 (m, 2H), 7.34 (d, J = 2.8 Hz, 1H), 7.09 (dd, J = 8.8, 2.8 Hz, 1H), 7.03 (d, J = 8.4 Hz, 2H), 6.56 (d, J = 7.4 Hz, 1H), 5.53 (s, 2H).

13

C NMR (126 MHz,

Chloroform-d) δ 162.31, 156.04, 155.81, 149.53, 148.93, 148.48, 135.09, 132.72, 131.91, 130.79 (2C), 128.61 (q, J = 31.6 Hz), 125.98, 125.61, 124.87, 124.12, 122.49, 122.35 (q, J = 273.9 Hz), 120.78, 119.30 (2C), 117.88 (q, J = 5.3 Hz), 114.98, 102.08, 68.27. HRMS (ESI): m/z [M+H]+ calculated for C25H17ClF3N4O2: 497.0987, found: 497.0988. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyridin-4-yl)imidazo[ 1,2-a]pyrimidine (10c). The title compound was obtained as a white solid from 9 and

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

4-bromopyridine using a method similar to that described for intermediate 4 in 48% yield. 1

H NMR (400 MHz, Chloroform-d) δ 8.74 (d, J = 6.0 Hz, 2H), 8.52 (d, J = 7.4 Hz, 1H),

7.81 (s, 1H), 7.54 (d, J = 8.4 Hz, 2H), 7.46−7.40 (m, 3H), 7.34 (d, J = 2.8 Hz, 1H), 7.09 (dd, J = 8.7, 2.8 Hz, 1H), 7.03 (d, J = 8.5 Hz, 2H), 6.60 (d, J = 7.4 Hz, 1H), 5.54 (s, 2H). 13

C NMR (126 MHz, Chloroform-d) δ 162.44, 156.07, 155.79, 150.80 (2C), 149.62,

136.37, 133.35, 133.16, 132.72, 131.79, 130.77 (2C), 129.60 (q, J = 31.8 Hz), 125.99, 122.52, 122.34 (q, J = 273.9 Hz), 121.56, 120.77 (2C), 119.31 (2C), 117.87 (q, J = 5.3 Hz), 102.32, 68.33. HRMS (ESI): m/z [M+H]+ calculated for C25H17ClF3N4O2: 497.0987, found: 497.0988. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(2-methoxypyrimidin5-yl)imidazo[1,2-a]pyrimidine (10d). The title compound was obtained as a white solid from 9 and 2-bromo-5-methoxypyrimidine using a method similar to that described for intermediate 4 in 43% yield. 1H NMR (300 MHz, Chloroform-d) δ 8.64 (s, 2H), 8.18 (d, J = 7.4 Hz, 1H), 7.62 (d, J = 8.7 Hz, 1H), 7.53 (d, J = 8.5 Hz, 2H), 7.43 (d, J = 8.7 Hz, 1H), 7.33 (d, J = 2.8 Hz, 1H), 7.08 (dd, J = 8.7, 2.8 Hz, 1H), 7.02 (d, J = 8.2 Hz, 2H), 6.57 (d, J = 7.1 Hz, 1H), 5.53 (s, 2H), 4.09 (s, 3H). 13C NMR (101 MHz, Chloroform-d) δ 165.40, 162.23, 158.50 (2C), 156.04, 155.85, 149.02, 132.73, 132.35, 132.34, 131.99, 130.69 (2C), 129.64 (q, J = 32.3 Hz), 126.00, 122.50, 122.36 (q, J = 274.5 Hz), 119.32 (2C), 117.89 (q, J = 5.4 Hz), 117.32, 117.04, 102.12, 68.20, 55.39. HRMS (ESI): m/z [M+H]+ calculated for C25H18ClF3N5O3: 528.1045, found: 528.1039.

ACS Paragon Plus Environment

Page 36 of 58

Page 37 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(1-methyl-1H-pyrazol -4-yl)imidazo[1,2-a]pyrimidine (10e). To a solution of 11 (30 mg, 0.06 mmol) and (1-methyl-1H-pyrazol-4-yl)boronic

acid

(16.3

mg,

0.078

mmol)

dissolved

in

dioxane-H2O (5 mL:1 mL) in a vial microwave tube, PdCl2(PPh3)2 (7.2 mg, 0.0012 mmol) and Na2CO3 (19.2 mg, 0.18 mmol) were added. The mixture was microwave irradiated at 150 °C for 0.5 h. After the mixture was cooled down, part of the solvent was evaporated under reduced pressure, the resulting solution was diluted with water, extracted with ethyl acetate (5 mL × 3), washed with brine, dried over NaSO4, filtered, and concentrated. The residue obtained was purified with preparative TLC (DCM/MeOH = 25:1) to yield 10e as a white solid (17 mg, 56%). 1H NMR (400 MHz, Chloroform-d) δ 8.21 (d, J = 7.4 Hz, 1H), 7.67 (s, 1H), 7.58 (s, 1H), 7.53 (d, J = 8.4 Hz, 2H), 7.49 (s, 1H), 7.44 (d, J = 8.8 Hz, 1H), 7.34 (d, J = 2.1 Hz, 1H), 7.08 (dd, J = 8.4, 2.1 Hz, 1H), 7.03 (d, J = 8.2 Hz, 2H), 6.50 (d, J = 7.4 Hz, 1H), 5.52 (s, 2H), 4.01 (s, 3H). 13C NMR (101 MHz, Chloroform-d) δ 161.74, 155.91, 155.89, 147.95, 138.09, 132.95, 132.70, 132.25, 130.85, 130.65 (2C), 129.59 (q, J = 32.1 Hz), 128.83, 125.89, 122.44, 122.36 (q, J = 274.4 Hz), 119.31 (2C), 117.84 (q, J = 5.4 Hz), 115.73, 109.37, 101.30, 67.96, 39.28. HRMS (ESI): m/z [M+H]+ calculated for C24H18ClF3N5O2: 500.1096, found: 500.1084. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-2-(pyrimidin-5-yl)imida zo[1,2-a]pyrimidine (10f). A solution of 8 (120 mg, 0.30 mmol) and crude 12 (60 mg) in ethanol (20 mL) was refluxed for 8 h. The solution was cooled down, diluted with

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 58

NaHCO3 solution, extracted with ethyl acetate (20 mL × 3), dried over Na2SO4, filtered, and concentrated. The residue obtained was purified with column chromatography (DCM/MeOH = 30:1) to yield 10f as a yellow solid (15mg, 10%). 1H NMR (300 MHz, Chloroform-d) δ 9.29 (s, 2H), 9.18 (s, 1H), 8.25 (d, J = 7.3 Hz, 1H), 7.76 (s, 1H), 7.52 (d, J = 8.5 Hz, 2H), 7.45 (d, J = 8.8 Hz, 1H), 7.35 (d, J = 2.8 Hz, 1H), 7.10 (dd, J = 8.9, 3.0 Hz, 1H), 7.05 (d, J = 8.6 Hz, 2H), 6.56 (d, J = 7.3 Hz, 1H), 5.55 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.86, 157.82, 156.11, 155.82, 153.92 (2C), 148.54, 139.18, 134.84, 132.74, 131.81, 130.58 (2C), 129.87 (q, J = 35.8 Hz), 127.49, 126.03, 122.53, 122.40 (q, J = 285.5 Hz), 119.35 (2C), 117.89 (q, J = 5.3 Hz), 106.68, 102.33, 68.34. HRMS (ESI): m/z [M+H]+ calculated for C24H16ClF3N5O2: 498.0939, found: 498.0948. 3-bromo-7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)imidazo[1,2-a]p yrimidine (11). To a solution of 9 (560 mg, 1.33 mmol) in THF (50 mL) was added NBS (262 mg, 1.33 mmol) at 0 °C, the reaction was stirred at rt for 1 h. After the reaction was complete, the solvent was evaporated under reduced pressure, the resulting residue was purified with column chromatography (petroleum ether/ethyl acetate = 4:1) to yield 11 as a yellow solid (240 mg, 36%). 1H NMR (400 MHz, Chloroform-d) δ 8.19 (d, J = 7.3 Hz, 1H), 7.52 (d, J = 8.6 Hz, 3H), 7.44 (d, J = 8.6 Hz, 1H), 7.34 (d, J = 2.7 Hz, 1H), 7.09 (dd, J = 8.8, 2.6 Hz, 1H), 7.03 (d, J = 8.4 Hz, 2H), 6.58 (d, J = 7.3 Hz, 1H), 5.51 (s, 2H). MS (ESI): m/z 498 [M+H]+. 2-bromo-1-(pyrimidin-5-yl)ethan-1-one

(12).

ACS Paragon Plus Environment

To

a

mixture

of

Page 39 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

1-(pyrimidin-5-yl)ethan-1-one (50 mg, 0.41 mmol), TMSOTf (225 µL, 1.23 mmol) and TEA (170 µL, 1.23 mmol) in THF (10 mL) was added NBS (80 mg, 0.45 mmol) at 0 °C, the mixture was stirred at 0 °C for 0.5 h. After the reaction was complete, the solution was diluted with water, extracted with ethyl acetate (10 mL × 3), dried over MgSO4, filtered, and concentrated. The resulting oil (60 mg) was used in next step without further purification. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)-3-methylbenzyl)oxy)-3-(pyrimidin-5 -yl)imidazo[1,2-a]pyrimidine (14a). The title compound was obtained as a white solid from 5 and 13a using a method similar to that described for compounds 7a-e in 38% yield. 1H NMR (400 MHz, Methanol-d4) δ 9.19 (s, 1H), 9.09 (s, 2H), 8.79 (d, J = 7.4 Hz, 1H), 7.78 (s, 1H), 7.53 (m, 2H), 7.42 (dd, J = 8.2, 1.8 Hz, 1H), 7.25 (d, J = 2.9 Hz, 1H), 7.06 (dd, J = 8.8, 2.9 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 6.72 (d, J = 7.4 Hz, 1H), 5.52 (s, 2H), 2.21 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 162.50, 157.97, 156.30, 155.04 (2C), 153.44, 149.66, 133.29, 132.64, 132.42, 132.35, 132.25, 130.40, 129.58 (q, J = 35.9 Hz), 127.97, 125.15, 123.79, 122.41 (q, J = 273.9 Hz), 120.90, 120.01, 117.30, 116.53 (q, J = 5.5 Hz), 102.51, 68.46, 16.13. HRMS (ESI): m/z [M+H]+ calculated for C25H18ClF3N5O2: 512.1096, found: 512.1126. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)-3-fluorobenzyl)oxy)-3-(pyrimidin-5yl)imidazo[1,2-a]pyrimidine (14b). The title compound was obtained as a white solid from 5 and 13b using a method similar to that described for compounds 7a-e in 75%

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

yield.

1

H NMR (300 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.93 (s, 2H), 8.35 (d, J = 7.4

Hz, 1H), 7.75 (s, 1H), 7.48−7.35 (m, 2H), 7.34−7.29 (m, 2H), 7.13 (t, J = 8.2 Hz, 1H), 7.05 (dd, J = 8.8, 2.5 Hz, 1H), 6.63 (d, J = 7.4 Hz, 1H), 5.56 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.20, 158.02, 155.88, 155.08 (2C), 153.11, 149.46, 142.246 (d, J = 11.8 Hz), 134.30 (d, J = 6.4 Hz), 133.38, 132.66, 132.56, 129.57 (q, J = 32.1 Hz), 125.92, 125.032 (d, J = 3.5 Hz), 123.72, 122.37, 122.32 (q, J = 273.9 Hz), 120.77, 117.55, 117.40, 116.33 (q, J = 5.4 Hz), 102.31, 67.57. HRMS (ESI): m/z [M+H]+ calculated for C24H15ClF4N5O2: 516.0845, found: 516.0831. 2-(4-chloro-3-(trifluoromethyl)phenoxy)-5-(((3-(pyrimidin-5-yl)imidazo[1,2-a]py rimidin-7-yl)oxy)methyl)benzonitrile (14c). The title compound was obtained as a yellow solid from 5 and 13c using a method similar to that described for compounds 7a-e in 64% yield.

1

H NMR (300 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.93 (s, 2H), 8.36 (d, J

= 7.4 Hz, 1H), 7.85 (d, J = 1.8 Hz, 1H), 7.75 (s, 1H), 7.71 (dd, J = 8.6, 2.0 Hz, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.42 (d, J = 2.8 Hz, 1H), 7.20 (dd, J = 8.7, 2.7 Hz, 1H), 6.94 (d, J = 8.6 Hz, 1H), 6.63 (d, J = 7.4 Hz, 1H), 5.56 (s, 2H). 13C NMR (101 MHz, Chloroform-d) δ 161.98, 158.07, 158.03, 155.11 (2C), 153.70, 149.29, 134.69, 133.95, 133.42, 133.23, 132.76, 132.42, 129.87 (q, J = 29.0 Hz), 128.20 (d, J = 1.7 Hz), 123.71, 123.65, 122.12 (q, J = 274.8 Hz), 118.47 (q, J = 5.4 Hz), 117.79, 117.51, 115.04, 104.82, 102.17, 66.87. HRMS (ESI): m/z [M+H]+ calculated for C25H15ClF3N6O2: 523.0892, found: 523.0868. 7-((3-chloro-4-(4-chloro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyrimidin-5

ACS Paragon Plus Environment

Page 40 of 58

Page 41 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

-yl)imidazo[1,2-a]pyrimidine (14d). The title compound was obtained as a white solid from 5 and 13d using a method similar to that described for compounds 7a-e in 36% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.94 (s, 2H), 8.37 (d, J = 7.4 Hz, 1H), 7.77 (s, 1H), 7.65 (d, J = 1.8 Hz, 1H), 7.44 (m, 2H), 7.29 (d, J = 2.9 Hz, 1H), 7.07 (d, J = 8.3 Hz, 1H), 7.01 (dd, J = 8.8, 2.8 Hz, 1H), 6.65 (d, J = 7.4 Hz, 1H), 5.55 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.21, 158.04, 155.53, 155.09 (2C), 150.97, 149.45, 134.01, 133.36, 132.72, 132.55, 131.07, 129.87 (q, J = 36.2 Hz), 128.47, 126.57, 126.05, 123.71, 122.34 (q, J = 268.1 Hz), 121.57, 121.20, 117.41, 116.78 (q, J = 5.3 Hz), 102.33, 67.48. HRMS (ESI): m/z [M+H]+ calculated for C24H15Cl2F3N5O2: 532.0549, found: 532.0539. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)-3-(trifluoromethyl)benzyl)oxy)-3-(p yrimidin-5-yl)imidazo[1,2-a]pyrimidine (14e). The title compound was obtained as a white solid from 5 and 13e using a method similar to that described for compounds 7a-e in 41% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.93 (s, 2H), 8.36 (d, J = 7.4 Hz, 1H), 7.85 (d, J = 1.7 Hz, 1H), 7.75 (s, 1H), 7.69 (dd, J = 8.5, 1.8 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 2.9 Hz, 1H), 7.11 (dd, J = 8.8, 2.8 Hz, 1H), 6.99 (d, J = 8.5 Hz, 1H), 6.63 (d, J = 7.4 Hz, 1H), 5.59 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.15, 158.06 (2C), 155.10 (2C), 153.89, 149.41, 133.83, 133.39, 132.95, 132.59, 132.03, 129.91 (q, J = 32.0 Hz), 127.75 (q, J = 4.8 Hz), 127.10, 123.69, 122.86, 122.86 (q, J = 273.3 Hz), 122.23 (q, J = 273.9 Hz), 122.33 (q, J = 30.9 Hz), 119.92, 118.26 (q, J

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

= 5.4 Hz), 117.44, 102.29, 67.49. HRMS (ESI): m/z [M+H]+ calculated for C25H15ClF6N5O2: 566.0813, found: 566.0830. 7-((4-(4-chloro-3-(trifluoromethyl)phenoxy)-3,5-difluorobenzyl)oxy)-3-(pyrimidi n-5-yl)imidazo[1,2-a]pyrimidine (14f). The title compound was obtained as a white solid from 5 and 13f using a method similar to that described for compounds 7a-e in 68% yield. 1H NMR (300 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.94 (s, 2H), 8.37 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.42 (d, J = 8.8 Hz, 1H), 7.30 (d, J = 2.9 Hz, 1H), 7.20 (d, J = 8.1 Hz, 2H), 7.03 (dd, J = 8.7, 2.7 Hz, 1H), 6.65 (d, J = 7.4 Hz, 1H), 5.56 (s, 2H). 13C NMR (101 MHz, Chloroform-d) δ 161.99, 158.10, 155.83, 155.71 (dd, J = 253.5, 4.4 Hz, 2C), 155.137 (2C), 149.29, 134.88 (t, J = 8.4 Hz), 133.40, 132.78, 132.61, 130.19, 129.53 (q, J = 32.2 Hz), 126.00, 123.63, 122.29 (q, J = 274.2 Hz), 119.36, 117.53, 115.10 (q, J = 5.4 Hz), 112.33 (dd, J = 17.3, 5.4 Hz, 2C), 102.18, 67.02. HRMS (ESI): m/z [M+H]+ calculated for C24H14ClF5N5O2: 534.0751, found: 534.0764. 7-((3,5-difluoro-4-phenoxybenzyl)oxy)-3-(pyrimidin-5-yl)imidazo[1,2-a]pyrimidi ne (14g). The title compound was obtained as a white solid from 5 and 13g using a method similar to that described for compounds 7a-e in 48% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.27 (s, 1H), 8.93 (s, 2H), 8.37 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.33−7.28 (m, 2H), 7.17 (d, J = 8.0 Hz, 2H), 7.07 (t, J = 7.4 Hz, 1H), 6.95 (d, J = 8.2 Hz, 2H), 6.65 (d, J = 7.4 Hz, 1H), 5.54 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.05, 158.05, 157.60, 156.14 (dd, J = 252.8, 4.8 Hz, 2C), 155.11 (2C), 149.36, 133.72 (t, J =

ACS Paragon Plus Environment

Page 42 of 58

Page 43 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

8.4 Hz), 133.39, 132.68, 131.151 (t, J = 15.4 Hz), 129.62 (2C), 123.68, 123.01, 117.46, 115.25 (2C), 112.17 (dd, J = 18.3, 5.3 Hz, 2C), 102.22, 67.20. HRMS (ESI): m/z [M+H]+ calculated for C23H16F2N5O2: 432.1267, found: 432.1267. 7-((3,5-difluoro-4-(4-fluoro-3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyrimidi n-5-yl)imidazo[1,2-a]pyrimidine (14h). The title compound was obtained as a white solid from 5 and 13h using a method similar to that described for compounds 7a-e in 63% yield. 1H NMR (300 MHz, Chloroform-d) δ 9.31 (s, 1H), 8.96 (s, 2H), 8.39 (d, J = 7.2 Hz, 1H), 7.81 (s, 1H), 7.24−7.08 (m, 5H), 6.73 (d, J = 7.3 Hz, 1H), 5.58 (s, 2H).

13

C NMR

(126 MHz, Chloroform-d) δ 161.96, 158.09, 155.77 (dd, J = 252.9, 4.2 Hz, 2C), 155.29 (d, J = 250.6 Hz), 155.12 (2C), 153.13, 149.30, 134.66 (t, J = 8.1 Hz), 133.43, 132.74, 130.64 (t, J = 15.1 Hz), 123.64, 122.03 (q, J = 273.0 Hz), 120.30 (d, J = 8.2 Hz), 118.11, 117.93, 117.50, 114.14 (d, J = 4.3 Hz), 112.32 (dd, J = 18.4, 4.8 Hz, 2C), 102.16, 67.02. HRMS (ESI): m/z [M+H]+ calculated for C24H14F6N5O2: 518.1046, found: 518.1105. 7-((3,5-difluoro-4-(3-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyrimidin-5-yl)im idazo[1,2-a]pyrimidine (14i). The title compound was obtained as a white solid from 5 and 13i using a method similar to that described for compounds 7a-e in 69% yield. 1H NMR (300 MHz, Chloroform-d) δ 9.30 (s, 1H), 8.95 (s, 2H), 8.39 (d, J = 6.5 Hz, 1H), 7.80 (s, 1H), 7.43 (t, J = 7.9 Hz, 1H), 7.34 (d, J = 7.8 Hz, 1H), 7.21 (d, J = 8.3 Hz, 3H), 7.12 (d, J = 7.1 Hz, 1H), 6.71 (d, J = 6.0 Hz, 1H), 5.58 (s, 2H).

13

C NMR (126 MHz,

Chloroform-d) δ 162.05, 158.09, 157.55, 155.85 (dd, J = 252.8, 4.7 Hz, 2C), 155.13 (2C),

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 44 of 58

149.30, 134.49 (t, J = 8.3 Hz), 133.32, 132.76, 132.16 (q, J = 32.9 Hz), 130.37 (t, J = 15.4 Hz), 130.29, 123.61, 123.58 (q, J = 279.1 Hz), 119.84 (d, J = 3.6 Hz), 118.53, 117.51, 112.45, 112.32 (dd, J = 19.0, 4.9 Hz, 2C), 102.25, 67.11. HRMS (ESI): m/z [M+H]+ calculated for C24H15F5N5O2: 500.1140, found: 500.1146. 7-((4-(3,4-dichlorophenoxy)-3,5-difluorobenzyl)oxy)-3-(pyrimidin-5-yl)imidazo[ 1,2-a]pyrimidine (14j). The title compound was obtained as a white solid from 5 and 13j using a method similar to that described for compounds 7a-e in 45% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.94 (s, 2H), 8.37 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.36 (d, J = 8.9 Hz, 1H), 7.19 (d, J = 8.0 Hz, 2H), 7.04 (d, J = 3.2 Hz, 1H), 6.84 (dd, J = 9.0, 2.8 Hz, 1H), 6.65 (d, J = 7.5 Hz, 1H), 5.56 (s, 2H).

13

C NMR (126 MHz,

Chloroform-d) δ 161.95, 158.06, 156.34, 155.76 (dd, J = 253.0, 4.4 Hz, 2C), 155.10 (2C), 149.29, 134.65(t, J = 8.1 Hz), 133.42, 133.20, 132.75, 130.93, 130.36 (t, J = 15.1 Hz), 126.62, 123.64, 117.49, 117.38, 115.11, 112.29 (dd, J = 18.4, 4.9 Hz, 2C), 102.15, 67.02. HRMS (ESI): m/z [M+H]+ calculated for C23H14Cl2F2N5O2: 500.0487, found: 500.0506. 7-((3,5-difluoro-4-(4-(trifluoromethyl)phenoxy)benzyl)oxy)-3-(pyrimidin-5-yl)im idazo[1,2-a]pyrimidine (14k). The title compound was obtained as a white solid from 5 and 13k using a method similar to that described for compounds 7a-e in 59% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.94 (s, 2H), 8.38 (d, J = 7.4 Hz, 1H), 7.77 (s, 1H), 7.57 (d, J = 8.6 Hz, 2H), 7.24−7.17 (m, 2H), 7.02 (d, J = 8.6 Hz, 2H), 6.66 (d, J = 7.4 Hz, 1H), 5.56 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 162.03, 159.77,

ACS Paragon Plus Environment

Page 45 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

158.12, 155.84 (dd, J = 253.0, 4.5 Hz, 2C), 155.15 (2C), 149.29, 134.60, 133.34, 132.75, 130.24 (t, J = 15.0 Hz), 127.17 (q, J = 3.6 Hz, 2C), 125.32 (q, J = 32.8 Hz), 124.03 (q, J = 271.9 Hz), 123.59, 117.52, 115.34 (2C), 112.26 (dd, J = 18.3, 4.8 Hz, 2C), 102.24, 67.09. HRMS (ESI): m/z [M+H]+ calculated for C24H15F5N5O2: 500.1140, found: 500.1170. 7-((4-((6-chloropyridin-3-yl)oxy)-3,5-difluorobenzyl)oxy)-3-(pyrimidin-5-yl)imid azo[1,2-a]pyrimidine (14l). The title compound was obtained as a white solid from 5 and 13l using a method similar to that described for compounds 7a-e in 34% yield.

1

H

NMR (400 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.94 (s, 2H), 8.37 (d, J = 7.4 Hz, 1H), 8.14 (d, J = 2.8 Hz, 1H), 7.76 (s, 1H), 7.26−7.24 (m, 2H), 7.20 (d, J = 8.1 Hz, 2H), 6.65 (d, J = 7.4 Hz, 1H), 5.56 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 161.91, 158.07, 155.54 (dd, J = 252.9, 4.3 Hz, 2C), 155.11 (2C), 153.19, 149.26, 145.00, 137.83, 134.94 (t, J = 8.2 Hz), 133.41, 132.79, 130.27 (t, J = 15.6 Hz), 125.72, 124.68, 123.62, 117.51, 112.32 (dd, J = 18.3, 4.8 Hz, 2C), 102.14, 66.94. HRMS (ESI): m/z [M+H]+ calculated for C22H14ClF2N6O2: 467.0829, found: 467.0849. 7-((3,5-difluoro-4-((6-(trifluoromethyl)pyridin-3-yl)oxy)benzyl)oxy)-3-(pyrimidi n-5-yl)imidazo[1,2-a]pyrimidine (14m). The title compound was obtained as a white solid from 5 and 13m using a method similar to that described for compounds 7a-e in 48% yield. 1H NMR (400 MHz, Chloroform-d) δ 9.28 (s, 1H), 8.93 (s, 2H), 8.49 (d, J = 2.7 Hz, 1H), 8.37 (d, J = 7.4 Hz, 1H), 7.76 (s, 1H), 7.64 (d, J = 8.6 Hz, 1H), 7.32 (dd, J = 8.9, 2.5

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Hz, 1H), 7.23 (d, J = 8.0 Hz, 2H), 6.65 (d, J = 7.4 Hz, 1H), 5.57 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 161.91, 158.13, 155.49 (dd, J = 253.3, 3.9 Hz, 2C), 155.48, 155.15 (2C), 149.3, 138.87, 135.5, 133.44, 132.78, 129.87 (q, J = 36.8 Hz), 128.64, 123.62, 122.50, 121.53 (d, J = 2.3 Hz), 120.33, 117.55, 112.37 (dd, J = 18.6, 4.8 Hz, 2C), 102.13, 66.91. HRMS (ESI): m/z [M+H]+ calculated for C23H14F5N6O2: 501.1093, found: 501.1132. ASSOCIATED CONTENT Supporting information Experimental procedures for intermediates 6a-e, 13a-m, HPLC for all the final compounds, and spectral data of compound 1. This material is available free of charge via the Internet at http://pubs.acs.org. AUTHER INFORMATION Corresponding Authors *Tel: 86-21-50806733 E-mail: [email protected]. *Tel: 86-21-20231969 E-mail: [email protected]. Author Contributions †

These authors contributed equally.

Notes

ACS Paragon Plus Environment

Page 46 of 58

Page 47 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

The authors declare no competing financial interest. ACKNOWLEDGMENTS This work was financially supported by grant from the National Science and Technology Major Project ‘Key New Drug Creation and Manufacturing program’ ( NOS. 2012ZX09103101-008). ABBREVIATIONS USED MW, molecular weight; cLogP, calculated logP; rt, room temperature; m-CPBA, 3-chloroperbenzoic

acid;

NBS,

bromosuccinimide;

TMSOTf,

trimethylsilyl

trifluoromethanesulfonate; TEA, triethylamine; DMA, N,N-dimethylacetamide; THF, tetrahydrofuran; DCM, dichloromethane; SD rats, Sprague-Dawley rats; TLC, thin-layer chromatography. REFERENCES 1.

Dennis, E. A.; Cao, J.; Hsu, Y. H.; Magrioti, V.; Kokotos, G. Phospholipase A2

enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev. 2011, 111, 6130-6185. 2.

Stafforini, D. M.; Elstad, M. R.; Mcintyre, T. M.; Zimmerman, G. A.; Prescott, S. M.

Human macrophages secrete platelet-activating factor acetylhydrolase. J. Biol. Chem. 1990, 265, 9682-9687. 3. Nakajima, K.; Murakami, M.; Yanoshita, R.; Samejima, Y.; Karasawa, K.; Setaka, M.;

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Nojima, S.; Kudo, I. Activated mast cells release extracellular type platelet-activating factor acetylhydrolase that contributes to autocrine inactivation of platelet-activating factor. J. Biol. Chem. 1997, 272, 19708-19713. 4.

Asano, K.; Okamoto, S.; Fukunaga, K.; Shiomi, T.; Mori, T.; Iwata, M.; Ikeda, Y.;

Yamaguchi, K. Cellular source(s) of platelet-activating-factor acetylhydrolase activity in plasma. Biochem. Biophys. Res. Commun. 1999, 261, 511-514. 5.

Caslake, M. J.; Packard, C. J.; Suckling, K. E.; Holmes, S. D.; Chamberlain, P.;

Macphee, C. H. Lipoprotein-associated phospholipase A2, platelet-activating factor acetylhydrolase: a potential new risk factor for coronary artery disease. Atherosclerosis 2000, 150, 413-419. 6.

MacPhee, C. H.; Moores, K. E.; Boyd, H. F.; Dhanak, D.; Ife, R. J.; Leach, C. A.;

Leake, D. S.; Milliner, K. J.; Patterson, R. A.; Suckling, K. E.; Tew, D. G.; Hickey, D. M. B. Lipoprotein-associated phospholipase A2, platelet-activating factor acetylhydrolase, generates two bioactive products during the oxidation of low-density lipoprotein: use of a novel inhibitor. Biochem. J. 1999, 338, 479-487. 7.

Shi, Y.; Zhang, P.; Zhang, L.; Osman, H.; Mohler, E. R.; Macphee, C.; Zalewski, A.;

Postle, A.; Wilensky, R. L. Role of lipoprotein-associated phospholipase A2 in leukocyte activation and inflammatory responses. Atherosclerosis 2007, 191, 54-62. 8.

Quinn, M. T.; Parthasarathy, S.; Steinberg, D. Lysophosphatidylcholine: a

chemotactic factor for human monocytes and its potential role in atherogenesis. Proc. Natl. Acad. Sci.U.S.A. 1988, 85, 2805-2809.

ACS Paragon Plus Environment

Page 48 of 58

Page 49 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

9.

Wilensky, R. L.; Macphee, C. H. Lipoprotein-associated phospholipase A2 and

atherosclerosis. Curr. Opin. Lipidol. 2009, 20, 415-420. 10. Lavi, S.; McConnell, J. P.; Rihal, C. S.; Prasad, A.; Mathew, V.; Lerman, L. O.; Lerman, A. Local production of lipoprotein-associated phospholipase A2 and lysophosphatidylcholine in the coronary circulation association with early coronary atherosclerosis and endothelial dysfunction in humans. Circulation 2007, 115, 2715-2721. 11. Racherla, S.; Arora, R. Utility of Lp-PLA2 in lipid-lowering therapy. Am. J. Ther. 2012, 19, 115-120. 12. Wilensky, R. L.; Shi, Y.; Mohler, E. R.; Hamamdzic, D.; Burgert, M. E.; Li, J.; Postle, A.; Fenning, R. S.; Bollinger, J. G.; Hoffman, B. E. Inhibition of lipoprotein-associated phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nat. Med. 2008, 14, 1059-1066. 13. Leach, C. A.; Hickey, D. M. B.; Ife, R. J.; Macphee, C. H.; Smith, S. A.; Tew, D. G. Lipoprotein-associated PLA2 inhibition—a novel, non-lipid lowering strategy for atherosclerosis therapy. Il. Farmaco. 2001, 56, 45-50. 14. Acharya, N. K.; Levin, E. C.; Clifford, P. M.; Han, M.; Tourtellotte, R.; Chamberlain, D.; Pollaro, M.; Coretti, N. J.; Kosciuk, M. C.; Nagele, E. P. Diabetes and hypercholesterolemia increase blood-brain barrier permeability and brain amyloid deposition: beneficial effects of the LpPLA2 inhibitor darapladib. J. Alzheimer’s Dis. 2013, 35, 179-198.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 50 of 58

15. Canning, P.; Glenn, J.; Prise, V.; Gale, D.; Stitt, A.; Adamson, P. Lp-PLA2 is a potential therapeutic target in diabetic macula edema. Invest. Ophthalmol. Vis. Sci. 2013, 54, 4613-4613. 16. Staurenghi, G.; Ye, L.; Magee, M. H.; Danis, R. P.; Wurzelmann, J.; Adamson, P.; McLaughlin, M. M. Darapladib, a lipoprotein-associated phospholipase A2 Inhibitor, in diabetic macular edema: a 3-month placebo-controlled study. Ophthalmology 2015, 122, 990-996. 17. Bresnick, G. H. Diabetic maculopathy: a critical review highlighting diffuse macular edema. Ophthalmology 1983, 90, 1301-1317. 18. Pendergast, S. D. Vitrectomy for diabetic macular edema associated with a taut premacular posterior hyaloid. Curr. Opin. Ophthalmol. 1998, 9, 71-75. 19. Tew, D. G.; Boyd, H. F.; Ashman, S.; Theobald, C.; Leach, C. A. Mechanism of inhibition of LDL phospholipase A2 by monocyclic-beta-lactams. Burst kinetics and the effect of stereochemistry. Biochemistry 1998, 37, 10087-10093. 20. Jeong, T. S.; Kim, M. J.; Yu, H.; Kim, K. S.; Choi, J. K.; Kim, S. S.; Lee, W. S. (E)-phenyl-

and

-heteroaryl-substituted

O-benzoyl-

(or

acyl)oximes

as

lipoprotein-associated phospholipase A2 inhibitors. Bioorg. Med. Chem. Lett. 2005, 15, 1525-1527. 21. Jeong, H. J.; Park, Y. D.; Park, H. Y.; Jeong, I. Y.; Jeong, T. S.; Lee, W. S. Potent inhibitors

of

lipoprotein-associated

phospholipase

A2:

benzaldehyde

O-heterocycle-4-carbonyloxime. Bioorg. Med. Chem. Lett. 2006, 16, 5576-5579.

ACS Paragon Plus Environment

Page 51 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

22. Lin, E. C. K.; Hu, Y.; Amantea, C. M.; Pham, L. M.; Cajica, J.; Okerberg, E.; Brown, H. E.; Fraser, A.; Du, L.; Kohno, Y.; Ishiyama, J.; Kozarich, J. W.; Shreder, K. R. Amides of xanthurenic acid as zinc-dependent inhibitors of Lp-PLA2. Bioorg. Med. Chem. Lett. 2011, 22, 868-871. 23. Hu, Y.; Lin, E. C. K.; Pham, L. M.; Cajica, J.; Amantea, C. M.; Okerberg, E.; Brown, H. E.; Fraser, A.; Du, L.; Kohno, Y.; Ishiyama, J.; Kozarich, J. W.; Shreder, K. R. Amides of 4-hydroxy-8-methanesulfonylamino-quinoline-2-carboxylic acid as zinc-dependent inhibitors of Lp-PLA2. Bioorg. Med. Chem. Lett. 2013, 23, 1553-1556. 24. Nagano, J. M. G.; Hsu, K. L.; Whitby, L. R.; Niphakis, M. J.; Speers, A. E.; Brown, S. J.; Spicer, T.; Fernandez-Vega, V.; Ferguson, J.; Hodder, P.; Srinivasan, P.; Gonzalez, T. D.; Rosen, H.; Bahnson, B. J.; Cravatt, B. F. Selective inhibitors and tailored activity probes for lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2013, 23, 839-843. 25. Boyd, H. F.; Flynn, S. T.; Hickey, D. M. B.; Ife, R. J.; Jones, M.; Leach, C. A.; Macphee, C. H.; Milliner, K. J.; Rawlings, D. A.; Slingsby, B. P.; Smith, S. A.; Stansfield, I. G.; Tew, D. G.; Theobald, C. J. 2-(alkylthio)pyrimidin-4-ones as novel, reversible inhibitors of lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2000, 10, 395-398. 26. Boyd, H. F.; Fell, S. C. M.; Flynn, S. T.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Macphee, C. H.; Milliner, K. J.; Moores, K. E.; Pinto, I. L.; Porter, R. A.; Rawlings, D. A.; Smith, S. A.; Stansfield, I. G.; Tew, D. G.; Theobald, C. J.; Whittaker, C. M. N-1

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 52 of 58

substituted pyrimidin-4-ones: novel, orally active inhibitors of lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2000, 10, 2557-2561. 27. Boyd, H. F.; Hammond, B.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Lewis, V. A.; Macphee, C. H.; Milliner, K. J.; Pinto, I. L.; Smith, S. A.; Stansfield, I. G.; Theobald, C. J.; Whittaker, C. M. The identification of a potent, water soluble inhibitor of lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2001, 11, 701-704. 28. Bloomer, J. C.; Boyd, H. F.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Macphee, C. H.; Milliner, K. J.; Pinto, I. L.; Rawlings, D. A.; Smith, S. A.; Stansfield, I. G.; Stanway, S.

J.;

Taylor,

M.

A.;

Theobald,

1-(arylpiperazinylamidoalkyl)-pyrimidones:

C.

J.;

orally

Whittaker,

active

C.

M.

inhibitors

of

lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2001, 11, 1925-1929. 29. Boyd, H. F.; Fell, S. C. M.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Macphee, C. H.; Milliner, K. J.; Pinto, I. L.; Rawlings, D. A.; Smith, S. A.; Stansfield, I. G.; Stanway, S. J.; Theobald, C. J.; Whittaker, C. M. Potent, orally active inhibitors of lipoprotein-associated phospholipase A2: 1-(biphenylmethylamidoalkyl)-pyrimidones. Bioorg. Med. Chem. Lett. 2002, 12, 51-55. 30. Blackie, J. A.; Bloomer, J. C.; Brown, M. J. B.; Cheng, H. Y.; Elliott, R. L.; Hammond, B.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Lewis, V. A.; Macphee, C. H.; Milliner, K. J.; Moores, K. E.; Pinto, I. L.; Smith, S. A.; Stansfield, I. G.; Stanway, S. J.; Taylor, M. A.; Theobald, C. J.; Whittaker, C. M. The discoverv of SB-435495: a potent, orally active inhibitor of lipoprotein-associated phospholipase A2 for evaluation in man.

ACS Paragon Plus Environment

Page 53 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Bioorg. Med. Chem. Lett. 2002, 12, 2603-2606. 31. Blackie, J. A.; Bloomer, J. C.; Brown, M. J. B.; Cheng, H. Y.; Hammond, B.; Hickey, D. M. B.; Ife, R. J.; Leach, C. A.; Lewis, V. A.; Macphee, C. H.; Milliner, K. J.; Moores, K. E.; Pinto, I. L.; Smith, S. A.; Stansfield, I. G.; Stanway, S. J.; Taylor, M. A.; Theobald, C. J. The identification of clinical candidate SB-480848: a potent inhibitor of lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 2003, 13, 1067-1070. 32. Jin, Y.; Wan, Z.; Zhang, Q. Compounds. US2012 / 0142717 A1, 2012. 33. Wan, Z.; Zhang, X.; Wang, J.; Peng, C.; Jin, Y.; Hu, Y. Compounds. WO2012 / 075917 A1, 2012. 34. Wan, Z.; Zhang, X.; Tong, Z.; Long, K.; Dowdell, S. E.; Manas, E. S.; Goodman, K. B. Tricyclic compounds, preparation methods, and their uses. WO2012 / 037782 A1, 2012. 35. Wan, Z.; Long, K.; Sang, Y.; Su, X. 2,3-dihydroimidazo[1,2-c]pyrimidin-5(1H)-one compounds use as Lp-PLA2 inhibitors. WO2013 / 013503 A1, 2013. 36. Wan, Z.; Long, K.; Zhang, X.; Yu, H. Bicyclic pyrimidone compounds. WO2013 / 014185 A1, 2013. 37. Wan, Z.; Zhang, X. Compounds. WO2014 / 114248 A1, 2014. 38. Wan, Z.; Zhang, X. Bicyclic pyrimidone compounds as inhibitors of Lp-PLA2. WO2014 / 114249 A1, 2014. 39. Sang, Y.; Wan, Z.; Zhang, Q. 2,3-dihydroimidazol[1,2-c]pyrimidin-5(1H)-one based lipoprotein-associated phospholipase A2 (Lp-PLA2) inhibitors. WO2014 / 114694 A1,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 54 of 58

2014. 40. Magrioti, V.; Kokotos, G. Phospholipase A2 inhibitors for the treatment of inflammatory diseases: a patent review (2010-present). Expert Opin. Ther. Pat. 2013, 23, 333-344. 41. White, H. D.; Held, C.; Stewart, R.; Tarka, E.; Brown, R.; Davies, R. Y.; Budaj, A.; Harrington, R. A.; Steg, P. G.; Ardis-Sino, D.; Armstrong, P. W.; Avezum, A.; Aylward, P. E.; Bryce, A.; Chen, H.; Chen, M. F.; Corbalan, R.; Dalby, A. J.; Danchin, N.; De Winter, R. J.; Denchev, S.; Diaz, R.; Elisaf, M.; Flather, M. D.; Goudev, A. R.; Granger, C. B.; Grinfeld, L.; Hochman, J. S.; Husted, S.; Kim, H. S.; Koenig, W.; Linhart, A.; Lonn, E.; Lopez-Sendon, J.; Manolis, A. J.; Mohler, E. R.; Nicolau, J. C.; Pais, P.; Parkhomenko, A.; Pedersen, T. R.; Pella, D.; Ramos-Corrales, M. A.; Ruda, M.; Sereg, M.; Siddique, S.; Sinnaeve, P.; Smith, P.; Sritara, P.; Swart, H. P.; Sy, R. G.; Teramoto, T.; Tse, H. F.; Watson, D.; Weaver, W. D.; Weiss, R.; Viigimaa, M.; Vinereanu, D.; Zhu, J. R.; Cannon, C. P.; Wallentin, L. Darapladib for preventing ischemic events in stable coronary heart disease. N. Engl. J. Med. 2014, 370, 1702-1711. 42. Mullard, A. GSK's darapladib failures dim hopes for anti-inflammatory heart drugs. Nat. Rev. Drug Discovery 2014, 13, 481-482. 43. Tawakol, A.; Singh, P.; Rudd, J. H. F.; Soffer, J.; Cai, G.; Vucic, E.; Brannan, S. P.; Tarka, E. A.; Shaddinger, B. C.; Sarov-Blat, L.; Matthews, P.; Subramanian, S.; Farkouh, M.; Fayad, Z. A. Effect of treatment for 12 weeks with rilapladib, a lipoprotein-associated phospholipase

A2

inhibitor,

on

arterial

inflammation

ACS Paragon Plus Environment

as

assessed

with

Page 55 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

F-18-fluorodeoxyglucose-positron emission tomography imaging. J. Am. Coll. Cardiol. 2014, 63, 86-88. 44. Wang, K.; Xu, W.; Liu, Y.; Zhang, W.; Wang, W.; Shen, J.; Wang, Y. Design and synthesis of imidazole and triazole derivatives as Lp-PLA2 inhibitors and the unexpected discovery of highly potent quaternary ammonium salts. Bioorg. Med. Chem. Lett. 2013, 23, 1187-1192. 45. Wang, K.; Xu, W.; Zhang, W.; Mo, M.; Wang, Y.; Shen, J. Triazole derivatives: A series of Darapladib analogues as orally active Lp-PLA2 inhibitors. Bioorg. Med. Chem. Lett. 2013, 23, 2897-2901. 46. Dave, M.; Nash, M.; Young, G. C.; Ellens, H.; Magee, M. H.; Roberts, A. D.; Taylor, M. A.; Greenhill, R. W.; Boyle, G. W. Disposition and metabolism of darapladib, a lipoprotein-associated phospholipase A2 inhibitor, in humans. Drug Metab. Dispos. 2013, 42, 415-430. 47. Molecular weight and cLogP value were calculated by ChemDraw 14. 48. Caprathe, B. W.; Jaen, J. C.; Wise, L. D.; Heffner, T. G.; Pugsley, T. A.; Meltzer, L. T.; Parvez,

M.

Dopamine

autoreceptor

agonists

as

potential

antipsychotics.

3.6-Propyl-4,5,5a,6,7,8-hexahydrothiazolo[4,5-f]quinolin-2-amine. J. Med. Chem. 1991, 34, 2736-2746. 49. Hu, Q.; Negri, M.; Jahn-Hoffmann, K.; Zhuang, Y.; Olgen, S.; Bartels, M.; Müller-Vieira, U.; Lauterbach, T.; Hartmann, R. W. Synthesis, biological evaluation, and molecular modeling studies of methylene imidazole substituted biaryls as inhibitors of

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 56 of 58

human 17α-hydroxylase-17,20-lyase (CYP17)—Part II: core rigidification and influence of substituents at the methylene bridge. Bioorg. Med. Chem. 2008, 16, 7715-7727. 50. Lucas, S.; Negri, M.; Heim, R.; Zimmer, C.; Hartmann, R. W. Fine-tuning the selectivity of aldosterone synthase inhibitors: structure−activity and structure−selectivity insights

from

studies

of

heteroaryl

substituted

1,2,5,6-tetrahydropyrrolo[3,2,1-ij]quinolin-4-one derivatives. J. Med. Chem. 2011, 54, 2307-2319. 51. Bischoff, F.; Berthelot, D.; De Cleyn, M.; Macdonald, G.; Minne, G.; Oehlrich, D.; Pieters, S.; Surkyn, M.; Trabanco, A. A.; Tresadern, G.; Van Brandt, S.; Velter, I.; Zaja, M.; Borghys, H.; Masungi, C.; Mercken, M.; Gijsen, H. J. M. Design and synthesis of a novel series of bicyclic heterocycles as potent γ-secretase modulators. J. Med. Chem. 2012, 55, 9089-9106. 52. Fu, R.; You, Q.; Yang, L.; Wu, W.; Jiang, C.; Xu, X. Design, synthesis and bioevaluation

of

dihydropyrazolo[3,4-b]pyridine

and

benzo[4,5]imidazo[1,2-a]

pyrimidine compounds as dual KSP and Aurora-A kinase inhibitors for anti-cancer agents. Bioorg. Med. Chem. 2010, 18, 8035-8043. 53. Al-Tel, T. H.; Al-Qawasmeh, R. A. Post Groebke-Blackburn multicomponent protocol:

synthesis

of

new

polyfunctional

imidazo[1,2-a]pyridine

and

imidazo[1,2-a]pyrimidine derivatives as potential antimicrobial agents. Eur. J. Med. Chem. 2010, 45, 5848-5855. 54. Steenackers, H. P. L.; Ermolat'ev, D. S.; Savaliya, B.; De Weerdt, A.; De Coster, D.;

ACS Paragon Plus Environment

Page 57 of 58

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Shah, A.; Van der Eycken, E. V.; De Vos, D. E.; Vanderleyden, J.; De Keersmaecker, S. C. J. Structure-activity relationship of 4(5)-aryl-2-amino-1H-imidazoles, N1-substituted 2-aminoimidazoles and imidazo[1,2-a]pyrimidinium salts as inhibitors of biofilm formation by Salmonella Typhimurium and Pseudomonas aeruginosa. J. Med. Chem. 2011, 54, 472-484. 55. Li, Q.; Woods, K. W.; Claiborne, A.; Gwaltney, S. L.; Barr, K. J.; Liu, G.; Gehrke, L.; Credo, R. B.; Hui, Y. H.; Lee, J.; Warner, R. B.; Kovar, P.; Nukkala, M. A.; Zielinski, N. A.; Tahir, S. K.; Fitzgerald, M.; Kim, K. H.; Marsh, K.; Frost, D.; Ng, S. C.; Rosenberg, S.; Sham, H. L. Synthesis and biological evaluation of 2-indolyloxazolines as a new class of tubulin polymerization inhibitors. Discovery of A-289099 as an orally active antitumor agent. Bioorg. Med. Chem. Lett. 2002, 12, 465-469. 56. Fu, H. Y.; Chen, L.; Doucet, H. Phosphine-free palladium-catalyzed direct arylation of imidazo[1,2-a]pyridines with aryl bromides at low catalyst loading. J. Org. Chem. 2012, 77, 4473-4478. 57. Benitez, S.; Sanchez-Quesada, J. L.; Ribas, V.; Jorba, O.; Blanco-Vaca, F.; Gonzalez-Sastre, F.; Ordonez-Llanos, J. Platelet-activating factor acetylhydrolase is mainly associated with electronegative low-density lipoprotein subfraction. Circulation 2003, 108, 92-96. 58. Samanta, U.; Bahnson, B. J. Crystal structure of human plasma platelet-activating factor acetylhydrolase: structural implication to lipoprotein binding and catalysis. J. Biol. Chem. 2008, 283, 31617-31624.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

59. Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47, 1739-1749.

Table of contents graphic

ACS Paragon Plus Environment

Page 58 of 58